• Shuffle
    Toggle On
    Toggle Off
  • Alphabetize
    Toggle On
    Toggle Off
  • Front First
    Toggle On
    Toggle Off
  • Both Sides
    Toggle On
    Toggle Off
  • Read
    Toggle On
    Toggle Off
Reading...
Front

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/204

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

204 Cards in this Set

  • Front
  • Back
What is the difference among:

Macrocytic Anemia

Normocytic normochromic Anemia

Microcytic hypochromic Anemia
What is the difference among:

Macrocytic Anemia- defective nuclear maturation; decrease production

Normocytic normochromic Anemia- Acute blood loss, hemolytic, renal failure, chronic disease

Microcytic hypochromic Anemia- Iron Deficiency
Name symptoms of Iron-Deficiency Anemia
Fatigue
SOB
Tachycardia
Koilonychia (spoon nails)
decreased cognitive function
Pica (craving to chew on ice, chalk, crayon or even dirt)
Name the glycoprotein that Iron is transfered by
Transferrin
What is the normal absorption of Iron?

What is the max?
Normal: 1-1.4 mg/day

Max: 3-4 mg/day

If you are iron deficient you are able to absorb more iron to compensate
What is the Iron requirement for:

Infant
Child
Adult male
Adult female
Pregnancy
What is the Iron requirement for:

Infant- 67 ug/kg
Child- 22 ug/kg
Adult male-13ug/kg
Adult female- 21 ug/kg
Pregnancy- 80 ug/kg
Foods High in Iron?

Foods Low in Iron?
High:
Meat
Brewer's Yeast
Wheat germ
Egg yolks
Oyster
Dried beans and fruit

Low:
Milk
Nongreen vegetables
Heme Iron vs. Inorganic Iron

which is more available?
which is more important?
Heme iron- more available

Inorganic iron- more important
Name 2 Facilitators of Iron Absorption
1. Ascorbic Acid (Vitamin C)
2. Meats (stimultes gastric acid)
Tolerable doses of oral iron in people with iron-deficiency anemia, will deliver at most how much iron/day to the erythroid marrow?
40-60 mg/day

increases RBC production by 2-3 times
Which is absorbed better?

Ferrous or Ferric Salts

Name some of theses
Ferrous are absorbed 3x better

Gluconate
Sulfate
Fumarate
Succinate
Name Ferrous Salts from highest percentage of Fe to least
FEOSTAT= 33% (sulfate)
FEOSOL= 20% (fumarate)
FERGON= 12% (gluconate)
NIFEREX= (succinate)
Name Absorption enhancers for Fe
increases absorption:

Ascorbic Acid (Vitamin C)
Carbohydrates
Amino Acids (meat)
inorganic salts
What are the RDA of iron for:

Pregnant females
lactating females
males
females >50
females 19-50
adolescent males >14
What are the RDA of iron for:

Pregnant females= 15mg, then 27 mg
lactating females= 9-10 mg
males= 8 mg
females >50= 8 mg
females 19-50= 18 mg
adolescent males >14= 11 mg
Oral Iron Doses for Iron Deficiency

Adults and Adolescents

Neonates, Infants, Children

Pregnant Women
Oral Iron Doses for Iron Deficiency

Adults and Adolescents- 200 mg/day

Neonates, Infants, Children- 3 mg/kg/ day

Pregnant Women- 15-30 mg/day
Oral Side Effects of Iron
Heartburn
Nausea
Upper gastric discomfort
Constipation and Diarrhea
What is Hemochromatosis?
Iron overload due to long-continued treatment (Iron Toxicity)

1-2 g in children (12-24months) can lead to death

Symptoms: abdominal pain, diarrhea, vomiting, cyanosis, lassitude, drowsiness, hyperventilation
Under what conditions do you consider Parenteral Iron?
Iron Malabsorption
Severe oral iron intolerance
Patients with renal disease who are receiving erythropoietin
What causes Megablastic Anemia?
deficiency in:

1. Vitamine B12
2. Folic Acid

results in defective synthesis of DNA
Name sources for Vitamin B12
Animal
Bacteria
How is Vitamin B12 absorbed?

What test is performed to evaluate the absorption of B12?

Is it okay to inject Cyanocobalamin I.V.?
from the ileum

complex of Intrinsic Factor B12

Intrinsic factor produced by parietal cells (this is why PPI are contraindicated)

Schilling test


NO- IM or SC only
Name some Therapeutic Uses of Vitamin B12
Trigeminal Neuralgia
Multiple Sclerosis
Neuropathies
Psychiatric disorders
What is the recommended daily requirement for Folic Acid?

What if pregnant or lactating?
400 ug

500-600 ug
What are some drugs that can cause Folate Deficiency?
anticonvulsants
methotrexate
trimethoprim
oral contraceptives

alcohol
A deficiency in what may cause spina bifida, encephaloceles, and anencephaly?
Folate Deficiency
Treatment for Folate Deficiency
Folic Acid (FOLVITE)

Folinic Acid (Leucovorin calcium, citrovorum factor)
Side Effects of high Folic Acid?
decreases antiepileptic actions of:

phenobarbital
phenytoin
primidone
What is the maximum level for Hemoglobin?

What are some Erythropoiesis Stimulating Agents (ESA)
Max: 12 g/L

Epoetin alfa
Darbepoetin alfa (Aranesp)
What diseases cause an increase in atherogenic risk?
coronary heart disease
Ischemic cerebrovascular disease
Peripheral vascular disease
Saturated fat should not take up more than what percent of total fat intake?
15%
What type of fatty acids increase levels of LDL cholesterol?
Trans fatty acids and saturated fatty acids

NOT unsaturated cis (olive oil)
Name the apoliproteins on these lipoproteins:

Chylomicrons

VLDL

LDL

HDL
Name the apoliproteins on these lipoproteins:

Chylomicrons- A, B48, C (i,ii, iii), E

VLDL- B100, C (i,ii,iii), E

LDL- B100

HDL- A, C (i,ii,iii), D, E
What is the precursor for LDL?
IDL
Which Lipoproteins do these functions?

Reverse cholesterol transport

Transport of endogenous triglycerides

Transport of dietary trigycerides

Major cholesterol transport lipoprotein
Which Lipoproteins do these functions?

HDL- Reverse cholesterol transport

VLDL- Transport of endogenous triglycerides

Chylomicron- Transport of dietary trigycerides

LDL- Major cholesterol transport lipoprotein
Lipoprotein Phenotype:

Type I
Type IIa
Type IIb
Type III
Type IV
Type V
Lipoprotein Phenotype:

Type I- deficiency in LPL
Type IIa- deficiency of LDL receptors
Type IIb- High Apo B & defective Apo E
Type III- abnormal IDL metabolism
Type IV- abnormal VLDL metabolism
Type V- abnormal VLDL and chylomicron metabolsim
Fiber
vs
Bile Acid Sequestrants
Fiber- decreases cholesterol absorption

Bile Acid Sequestrants- decrease bile acid reabsorption
What is the enterohepatic circulation?
cycle of secretion adn reabsorption of bile salts and cholesterol
stored in the gall bladder, amphipathic, and acts as detergent to break up dietary lipids

transfered back to liver bound to albumin
Bile Salts
Reduces cholesterol adsorbtion in to the blood stream

binds bile salts and cholesterol and prevent its reabsorption
Sitostanol ester

Fiber
group of medication used for binding bile in the gastrointestinal tract; treats hypercholesterolemia and chronic diarrhea
Bile Acid Sequestrants: Resins
What is the mechanism of action for bile acid sequestrant?
Anion-exhange resins (binds anions)

increases bile acid synthesis
stimulates production of LDL receptors
lowers LDL-C levles
increases LDL clearance
increase in TG production
Name 3 Bile Acid Sequestrants
1. Cholestyramin (Questran)
2. Colestipol (colestid)
3. Colesevelam (WelChol)

Cholestyram and Colestipol are more effective at lowering LDL, while Colesevelam is more effective at raising HDL; both may increase TG
Drug interactions with colestipol and cholestyramine?
binds to other negatively charged drugs

must give other drugs 1 hour before or 4-6 hours after

interfere with the absorption of folic acid and fat-soluble vitamines
What are some of the uses of Bile Acid Sequestrants?
1. Adjunct to treatment of hypercholesterolemia
2. Pruritus due to high bile acids; diarrhea associated with excess fecal bile acids
3. Binding toxicologic agents
4. Oxaluria- excess calcium oxalae in the urine
Which drugs do Bile Acid Sequestrants interact with?
Some thiazides
Furosemide
Propranolol
L-thyroxine
Cardiac glycosides
Coumadin anticoagulants
Some statins
Statins Mechanism of Action
inhibition of HMG-CoA reductase
increase LDL receptors in liver
increase LDL clearance
What are the LDL levels for:

Optimal
Near optimal
Borderline High
High
Very High
What are the LDL levels for:

Optimal- <100 mg/dl
Near optimal 100-129
Borderline High 130-159
High 160-180
Very High >190
Total Cholesterol Levels

Desirable
Borderline High
High
Total Cholesterol Levels

Desirable <200 mg/dl
Borderline High 200-239 mg/dl
High >240 mg/dl
HDL Cholesterol Levels

Low

High
HDL Cholesterol Levels

Low < 40 mg/dl (bad)

High >60 mg/dl (good)
Name the Equivalent Diseases to Coronary Heart Disease
CHD
Carotid Artery Disease
Peripheral Artery Disease
Abdominal Aortic Aneurysm
Name 5 Risk Factors for Hypercholesteremia
1. Cigarette Smoking
2. Hypertension (>140/90)
3. Low HDL (<40 mg/dl)
4. Family History of CHD
5. Age: Men 45 Women 55
What are the LDL-Goal and levels at which to consider drug therapy for:

CHD or CHD equivalent

>2 risk factors (10-20%)

>2 risk factors (<10%)

<2 risk factors
What are the LDL-Goal and levels at which to consider drug therapy for:

CHD or CHD equivalent: <100/ >130

>2 risk factors (10-20%): <130/ >130

>2 risk factors (<10%): <130/ >160

<2 risk factors: <160/ >190
Name of Calculator for estimating 10-year CHD Risk
Framingham
Name the 6 Statins with their strengths that are dose equivalent (most potent to least)
Rosuvastatin 5mg
Atorvastatin 10 mg
Simvastatin 20mg
Lovastatin 40 mg
Pravastatin 40 mg
Fluvastatin 80mg
When is someone considered to have Metabolic Syndrome?
when > or = 3 risk factors are present

Waist:
male= >40in
female= >35

TG: >150 mg/dl

HDL:
men= <40
women= <50

Blood pressure: >130/85

Fasting glucose: >110 mg/dl
Triglyceride levels for:

Normal

Borderline High

High

Very High
Triglyceride levels for:

Normal <200

Borderline High 200-399

High 400-1000

Very High >1000
Compare LDL cholesterol and Non-HDL Cholesterol goals for:

CHD equivalent

>2 Risk Factors

0-1 Risk Factor
Compare LDL cholesterol and Non-HDL Cholesterol goals for:


LDL-goal/ Non-HDL goal

CHD equivalent: <100/ <130

>2 Risk Factors: <130/ <160

0-1 Risk Factor: <160/ <190
No atherosclerosis progress when LDL is below what?
<67 mg/dl
Is Diabetes a CHD equivalent?
YES
What is the most significant side effect of Statins?
Myopathy and Rhabdomyolysis
Doubling the dose of statins will increase the effect by how much?
~5-6%
Steps of Plaque formation?
1. LDL enters artery wall
2. Modified LDL stimulates expression of MCP-1 in endothelial cells
3. Monocytes differentiate into Macrophages
4. Modified LDL indudes Macrophages to release cytokines that stimulate Adhesion molecule expression in endothelial cells
5. Macrophages express receptors that take up modified LDL
6. Macrophages and Foam cells express Growth Factors and Proteinases
7. remnants of VLDL and Chylomicrons are also proinflammatory
Where is HDL produced?
Liver and Intestine
What is involved in the HDL metabolism and reverse cholesterol transport?
Nascent HDL picks up FC from macrophage using ABC1

Nascent HDL changes to mature HDL using LCAT

Mature HDL with CE now binds SR-BI on Liver
HMG-CoA Reductase Inhibitors
(Differences in effect on LDL/ HDL/ TG)

Rosuvastatin
Atorvastatin
Simvastatin
Lovastatin
Pravastatin
Fluvastatin
HMG-CoA Reductase Inhibitors
(Differences in effect on LDL/ HDL/ TG)

Rosuvastatin +++/+++/++
Atorvastatin ++/+/+
Simvastatin ++/++/+++
Lovastatin ++/++/++
Pravastatin +/+/+
Fluvastatin +/+/+
Statin Combos:

Statin-bile acid

Statin-Niacin

Statin-Fibrate
Statin Combos:

Statin-bile acid- 20-30% more reduction in LDL-C

Statin-Niacin- increases risk of myopathy

Statin-Fibrate- useful in patients with both high TG and LDL-C

Triple Therapy= up to 70% reduction in LDL-C
Four mechanisms by which Statins are thought to prevent cardiovascular diease
1. Improve endothelial function
2. Modulate inflammatory responses
3. Maintain plaque stability
4. Prevent Thrombus formation
What is Myopathy?
major adverse effect of statins

skeletal inhibition of skeletal muscle sterol synthesis
Drug interactions with Statins?
Macrolides
antifungals
cyclosporine
phenylpiperazine antidepressants
nefazodone
pretease inhibitors
amiodaron
warfarin

Both use CYP3A4
Which statins are water soluble and which are lipophilic?
Lipophilic:
Atorvastatin
Simvastatin
Lovastatin
Fluvastatin

Water soluble:
Rosuvastatin
Pravastatin
Statins the use CYP2C9 and can interact with:

Amiodarone
Gemfibrozil
Fluconazol
metronidazole
Fluoxetine
Warfarin
Fluvastatin
and
Rosuvastatin
Which Statin is associated with an increased risk of muscle injury?
Simvastatin 80mg
Can Statins be used in Pregnancy?
NO

Risk Factor X
Foods and/or products to avoid when taking statins?
Ethanol (+)
Grapefruit juice (+)
St John's wort (-)
How do Fibrates work?
Fibrates activate PPARs causing transcription of a number of genes on the DNA that facilitate lipid metabolism

They are Amphipathic carboxylic acids
What types of elevated TG do Fribrates treat?
Type III- IDL
Type IV- high VLDL
Type V- high VLDL and chylomicron
Name 2 Fibrates
1. Gemfibrozil (Lopid)
2. Fenofibrate (Antara, Lofibra, TriCor, Triglide)
What is the active metabolite of Fenofibrate?
Fenofibrate is a prodrug

active metabolite- fenofibric acid
Side Effects of Fibrates
Stomach upset
Myopathy
Rhabdomylysis
Lithiasis (predisposition to the formation of gallstones)
Drug Interactions with Fibrates
Warfarin
What is the importance of Niacin? and what are other names for Niacin?
Nicotinic acid and Vitamine B3

water soluble vitamin that playes role in energy metabolism

deficiency in Niacin leads to Pellagra and Depression
Tryptophan is used to synthesize Niacin and Serotonin

therefore, niacin deficiency can cause what?
Depression b/c lack of serotonin
What are the actions of Niacin
Inhibits VLDL secretion
Inhibits free fatty acid release
Increases LPL activity
Decreases TG synthesis and lipolysis
Reduces TG
Side Effects of Niacin?
1. Liver Toxicity
2. Flushing

take Aspirin 30 min prior to help prevent flushing

the flushing is harmless

also could try suspended release instead of immediate release
Name 2 Dietary and Biliary Cholesterol Absorption Inhibitors (blocks intestinal absorption of cholesterol)
1. Ezetimibe (Zetia) 19% decrease in LDL-C
2. Vytorin (simvastatin with acetamide) 52% decrease in LDL-C
Name 3 combination drugs for hypercholesteremia
1. Advicor (Niacin and Lovastatin)- take at bedtime; lowers LDL, TG, VLDL, and blocks cholesterol synthesis

2. Simcor (Niacin ER and Simvastatin)

3. Caduet (Atorvastatin and Amlodipine) blocks cholesterol synthesis and lowers blood pressure
Name potential Triglyceride-Lowering Mechanisms of Omega-3 Fatty Acids
1. Decrease in lipogenesis
2. Increase in Beta-oxidation, phospholipid synthesis, and apo B degradation
3. reduced secretion of VLDL and TG
What is in Lovaza?
DHA
EPA
Omega-3
What is the formula for the omega-3 Index?

Is a higher or lower number better?
(EPA+DHA)/ Total Fatty Acids

You want a high % by eating more fish
What are some of omega-3 Drug interactions?
- Blood Thinning medications (warfain, aspirin, clopidogrel)

- Blood Sugar Lowering medications: increases blood sugar levels

Beneficial for:

Cyclosporine, Cholesterol-lowering medications, NSAIDS

NSAIDS with omega-3 decreases ulcers
What levels can be measured at non-fasting state?

Apo B/ Apo A-I should be less than what?

Total C/ HDL should be less than what?
Total Cholesterol
and
HDL

Apo B (LDL)/ Apo A-I (HDL) <0.9

Total C/ HDL <4.5
What is the spontaneous cessation of blood loss from a damaged blood vessel
Hemostasis
Three main processes of Hemostasis
1. Adhesion and activation of platelets
2. Fibrin formation (blood coagulation)
3. Vascular contraction of damaged blood vessels
What is the removal of a clot called?
Fibrinolysis
Explain the process that occurs after PGI2 binds to a platelet
PGI2 binds to platelet

increase in cAMP

Ca2+ sequation inhibiting platelet aggregation
due to exposure to collagen?
ADP
5HT
TXA

causes aggregation
Name platelet signals that are activators
ADP
Collagen
Thrombin
Thromboxanes
Serotonin
Epinephrine
Antigen-Antibody Complex
Viruses and Bacteria
Name platelet signals that are inhibitors
Prostacyclin (PGI2)
cAMP
Decreased levels of thrombin and thromboxanes
What does blood coagulation do?
transforms soluble fibrinogen into insoluble fibers
Enzyme that converts fibrinogen to fibrin
Thrombin
Thrombin activates what to further stabalize clots?
Crosslinking action of Factor XIII
Which is the dominant pathway in the body?

Intrinsic (tissue not in contact with blood)
or
Extrinsic (injured tissue contact with blood)
Intrinsic
What does the Intrinsic Pathway require besides IX and VII to make IXa and VIIa

also needed for production of Xa from A
Ca2+
Phospholipid
VIIIa
IIa is called what?

I is called what?

II is called what?
IIa- Thrombin

I- Fibrinogen

II- Prothrombin
Coagulation proteins are inactivated by...
alpha2- macroglobin

alpha2- antiplasmin

Antithrombin III; Protein C
Proteins C and S inactivate which factors?
Factor Va

Factor VIIIa
What is Fibrinolysis?
conversion of inactive plasminogen to plasmin

Plasmin digests fibrin to limit the extent of thrombosis
clot that adheres to a vessel wall

clot that floats within the blood
Thrombus

Embolus
White clot vs. Red clot
White- found in rapidly flowing (arterial) blood; treated with antiplatelet drugs and anticoagulants

Red- found in slow moving (venous) or stagnant blood; treated with anticoagulants and fibrinolytic drugs
If clot forms in leg it is known as what?

If clot forms in lungs, it is known as what?

If clot forms in the heart, it is known as what?
Deep Vein Thrombosis

Pulmonary Embolism

Heart Attack
Name some anticoagulants and what do these drugs affect?
Argatobran
Lepirudin
Warfarin

taregets anticoagulant factors
Name antiplatelets and what they target
Aspirin
Ticlopidine (irreversible)
Clopidogrel (irreversible)
Dipyridamole
Abciximab
Eptifibatide
Tirofiban

Targets platelets
Name some Thrombolytics and what do they target?
Streptokinase
Anysoylated

Targets plasminogen
Name some Antithrombotic agents and what do they target
Aminocaproic acid
Tranexamic acid
Aprotinin
What is Antithrombin III (ATIII)
natural serine protease inhibitor that inhibits coagulation factors:
IXa
Xa
IIa (Thrombin)

by binding to their active sites
How does Heparin Anticoagulate
Heparin molecules bind ATIII inducing a conformation change that accelerates its rate of action about 1000-fold (inhibiting coagulation factors IXa, Xa, IIa)
Difference between Regular Heparin and LMW Heparin
Regular- blocks IXa, Xa, IIa, by binding to IIa and/or Xa

LMW- only inhibits Xa
What is the drug of choice for anticoagulation during Pregnancy?
Heparin- cannot cross placenta
What do you measure for monitoring parameters in patients taking Heparin?
aPTT (Activated partial thromboplastin time)

should be 1.5-2x patients' baseline measures intrinsic pathway
Heparin is contraindicated in Patients that are what?
bleeding
have hemophilia
thrombocytopenia
hypertension

Do NOT take with Aspirin or positively charged drugs and aminoglycosides
Name 3 LMWH

LMW Heparins

What is the target for these drugs?
1. Enoxaparin (Lovenox)
2. Dalteparin (Fragmin)
3. Tinzaparin (Innnohep)

Potent factor Xa inhibitor

Given SC
How is the biological activity of LMW Heparins measured?
Factor Xa inhibition assay

less monitoring than heparin

No aPTT needed

Monitor end stage renal disease
What drugs inhibits thrombin-factor IIa
Lepirudin (Refludan)- Hirudin derivative from saliva of medicinal leeches; administered IV, aPTT monitorring recommended

Argatroban- side effect is bleeding
Name the Parenteral Anticoagulant that is purely synthetic derived and no variable biological activity
Fondaparinux (ARIXTRA)

Selectively inhibits factor Xa by binding to ATIII

well absorbed from SC route

contraindicated with patients with severe renal impairment

does not affect platelet function or actions
No HIT (Heparin induced thromboyctopenia)
How is Warfarin administered and how does it work?
Warfarin (Coumadin) is an oral anticoagulant

antagonist of vitamin K-cofactor in the hepatic synthesis of coagulation factors II, VII, IX, X and antocoagulant proteins C and S

blocks reduction of oxidized vitamin K therefore blocking carboxylation

targets synthesis of coagulation factors

*Vitamin K epoxide Reductase*
Which anticoagulant has 100% bioavailability?
Warfarin (Coumadin)
Onset of action for Warfarin depends on the dissapearance of which factors?
VII, IX, X, II

Peak effects may not be until 72-96 hours (3-5 days) which is the time required to deplete the pool of circulating clotting factors
Name some Indicatications for the use of Warfarin
-prevent progression of acute deep vein thrombosis or pulmonary embolism

-prevention of venous thromboembolism during orthopedic or gynecological surgery

- Prophylaxis in patients with acute MI, prosthetic heart valves, or chronic atrial fibrillation
What are Side Effects of Warfarin?
Bleeding
skin lesions and skin necrosis
purple toe syndrome
What drugs are included in Drug interactions with Warfarin
Aspirin and phenylbutazone (inc. bleeding)

Antibiotics (decreases metabolsim of warfarin)

Barbituates and Rifampin (decrease warfarin effectiveness by inducing microsomal P450 system)

-Oral contraceptives (dec warfarin effect by inc. plasma clotting factors and decreasing ATIII)
What drugs decrease the anticoagulant effects of Warfarin
Estrogen
Phenytoin
Rifampin
Barbiturates
Vitamin K
What drugs increase the bleeding tendencies with Warfarin?
Cephalosporins
Salicylates
Adrenal corticosteroids
NSAIDS
What drugs enhance anticoagulant effects of Warfarin?
Oral antibiotics
Salicylates
Alcohol
Sulfonamides
Cimetidine
Amiodarone
How is Warfarin monitored?

What is the goal?
Prothrombin time (PT)

International Normalized Ratio (INR)

Goal: INR= 2-3 or INR= 2.5-3.5 (mechanical heart valves)
When should Warfarin be discontinued?

How is Warfarin toxicity treated?
Discontinue if INR <5

administer large amount of Vitamin K (phytonatoide) if INR >5

serious bleeding with INR> 20 requires vitamin K, factor IX concentrates and fresh frozen plasma to replace clotting factors
Heparin vs Warfarin

Route
Onset
Duration
Monitoring
Antidoe for overdose
Heparin vs Warfarin

Route IV or SC/ PO
Onset Rapid/ Slow
Duration Brief/ Prolonged
Monitoring aPTT/ PT
Antidoe for overdose Protamine/ Vitamin K
Oral anticoagulant that is a direct thrombine inhibitor
Dabigatran (Pradaxa)

approved in Europe

no monitoring
Inhibits synthesis of prostaglandins by decreasing endothelial synthesis of PGI2 and inhibitibiting the production of TXA2 by acetylating the active site of cyclooxygenase
Aspirin
Uses for Aspirin
Prophylactic for transient cerebral ischemia

Reduce the incidence of recurrent MI

Decrease mortality in post-myocardial infarction patients
Agents that irreversibly inhibit the binding of ADP to its receptor on platelets?
1. Ticlopidine (prodrug)
2. Clopidogrel

target is ADP
nucleotide that causes platelets to change shape and aggregate
ADP
Ticlopidine

Clopidogrel
Ticlopidine- more severe side effects like nausea, vomiting, and diarrhea; NEUTROPENIA is most serious side effect; THROMBOCYTOPENIA also reported

Clopidogrel- prodrug converted via CYP450 enzyme; more favorable toxicity profile

Both are prodrug
Black Box Warning for Clopidogrel
CYP2C19 activation required by clopidogrel to convert it's active metabolite

higher cardiovascular events with patients with poor CYP2C19 metabolism
Direct acting oral ADP receptor inhibitor not requiring hepatic metabolism for activation
Ticagrelor (Brilinta)

not approved in USA yet

faster onset, more potent, short half-life, reversible

greater incidence of dyspnea, ventricular pauses and increased levels of creatinine
vasodilator that inhibits platelet aggregation by increasing cellular concentration of cyclic AMP
Dipyridamole (Persantine)
Antiplatelet Agents that prevent binding of fibrinogen, vWF, and other adhesive molecules to GP IIb/IIIa receptor sites

Which one is derived from rattle snake venom

which one is a fragment of humanized monoclonal antibody?
Abciximab (Reopro)- antibody

Tirofiban (Aggrastat)

Eptifibatide (Integrilin)- derived from rattle snake venom

both inhibit GPIIb/IIIa receptor and are IV administered
How do Thrombolytic agents work?

Name two
Anistreplase (Eminase)

Altepase

activate plasminogen and dissolve both pathological thrombi and fibrin deposits at vascular sites

venous thrombi are easier to lyse than arterial thrombi

primarily used to dissolve clots in patients
nonspecific serine protease that digests fibrin and other plasma proteins including several coagulation factors
Plasmin
What is the endogenous inhibitor of plasmin
alpha2-antiplasmin
Thrombolytic Agent that is Anysoylated Plasminogen Activator Complex

what is the purpose of this?
Anistreplase (Eminase)

protects streptokinase from hydrolysis when it is bound to plasminogen

targets fibrinolytic clots
Thrombolytic Agent that is a tissue plasminogen activator (t-PA)

What is the purpose of this?
Altepase

activates plasminogen bound to fibrin several hundred times more rapidly than unbound plasminogen

used to lyse thrombi
Name the thrombolytic agents

recombinant forms of human tissue plasminogen activator (t-PA)

a protein obtained from human urine

protein obtainedfrom streptococci

preformed complex of streptokinase and plasminogen-prodrug
Alteplase (Activase)

Urokinase (Abbokinase)

Streptokinase (Streptase)

Anistreplase (Eminase)
Which thrombolytic agents directly convert plasminogen to plasmin

and

which must combine with plasminogen first

which must be released from plasminogen
Urokinase and t-PA

Streptokinase

Anistreplase and Streptokinase
Major side effect of Thrombolytic Agents
Hemorrhage-intracranial bleeding

can be controlled by aminocaproic acid
Contraindications with Thrombolytic Agents
Surgery within 10 days
Serious GI bleeding
Hypertension history
active bleeding
kinetic screening test that measures the recalcification time of plasma after incubation with phospholipid and kaolin using a semi-automated method (instrinsic system)

used to evaluate the adequacy of the extrinsic system
APTT
Activated partial thromboplastin Time

PT- Prothrombine Time
measures the clotting ability for factors I, II, V, VIII, X
Repolarization is mainly determined by what?
openings of the potassium channels, leading to potassium exit from the cells
What is special about the pacemaker cells?

Which is the dominant pacemaker in the heart?
spontaneous phase 4 depolarization

SA node is dominant over the AV node
Which class of drugs depresses the SA and AV potentials?
CCB
What is the Effective refractory period (ERP)?
time elapsed from initiation of the action potential to the moment where a stimulus elicits a conducted response
What determines the conduction velocity of the cell?
phase 0 of the action potential (sodium entry)

steeper=faster conduction velocity
His-Purkinje/myocardial cells
vs
Atrial cells
His-Purkinje/myocardial cells- large sodium currents with large action potentials (no pacemaker activity)

Atrial Cells- shorter action potentials due to stronger repolarizing currents
Voltage-dependent potassium channels play a role in what phases?

K influx or K efflux?

Potassium channels determine what about the action potential?
Phases 2,3,4

K efflux

duration
What does blocking the potassium currents do?
causes slower repolarization

prolongs action potentials
Drugs that inhibit potassium currents
Bretylium
Amiodarone
Sotalol
Quinidine
Procainamide
Dispyramide

antibiotics and antidepressants
Voltage-dependent calcium channels are found in which phases? (efflux or influx)

what are the main type of calcium channels in the heart tissue?
phase 2 (plateau)

Ca2+ influx

L-type channels
L-type calcium channels are sensitive to which drugs?
DHP CCB and non DHP CCB
What protein does Calcium bind to once it goes back into the sarcoplasmic reticulum?
Calsequestrin
through what methods does the Ca leave the cell?
sodium-calcium exchange
(2 Ca ou and 3 Na in0

Ca-ATPase pummp

taken up by sarcoplasmic reticulum
By decreasing calcium entry, CCBs are expected to decrease what?
decrease contractility
In SA and AV nodes, depolarization is mainly due to what?
calcium inward currents (little sodium influx0

CCB depress SA and AV potentials reducing Hr and conduction velocity through the AV node
Sodium-Potassium ATPase exchanges what and where?

Drug that inhibits it
3 Na out; 2 K in

Digoxin

augments strength of contraction because high level of Na will trigger the Na/Ca exchanger to let Na out and bring Ca in
What happens during each phase?

P wave:
P-R interval:
QRS complex:
ST segment:
T wave:
QT interval:
What happens during each phase?

P wave: atrial depolarization
P-R interval: atrial repolarization?
QRS complex:ventricular depolarization
ST segment: beginning of repolarization
T wave: Ventricular Repolarization
QT interval:
Range for normal resting HR

Pacemaker site?
60-100 b/min

SA node
___________ reflects the capability of a cardiac cell to depolarize spontaneously during phase 4 to reach threshold potential and to depolarize completely without being externally stimulated
Automaticity
Capability of a resting, polarized cardiac cell to depolarize in response to an electrical stimulus. All cardiac cells have this capability
Excitability
Mechaisms by which arrhythmias are produced?
1. Increased automaticity
2. Afterdepolarizations
3. Reentry
Increased automacticity can be caused by what?
SNS activation
Beta stimulation
Low potassium
Fiber stretch
Inhibition parasympathetic acitivity
anticholinergic drugs

OR

increased automacticity in non-pacemaker cells due to ischemia of ventrcular cells or anti-arrhythmic drugs
Drugs that prolong the QT interval
Antimicrobials:
macrolides (erythromycin, chlarithromycin)
Pentamidine
Fluoroquinolones (grepafloxacin, moxifloxacin, levofloxacin)

Tricyclic antidepressants:
imipramine
desipramine
amitrypiline
doxepin

Thioridazine, mesoridazine, haloperidol, resperidone, ziprasidone, quetiapine
Cisapride
Indapamide
How do drugs like azole antifungul agents prolong the QT interval?
block the metabolsim of terfenadine
What is the difference between EAD and DAD?
Early afterdepolarization (EAD) occurs during phase 3- determined by calcium currents

Delayed afterdepolarization (DAD) arise after action potential has completed- sodium and calcium currents determine the upstroke of DAD
AV nodal block can be achieved by what?
increased vagal tone
adenosine
verapamil
diltiazem
beta adrenergic blockers
digitalis
What are the classifications for anti-arrhythmic drugs?
Type 1: Sodium Channel Blockers
Type 2: Beta Blockers
Type 3: Potassium channel blockers
Type 4: Calcium Channel Blockers
Names for type 1 anti-arrhythmic sodium channel blockers

IA
IB
IC
Names for type 1 anti-arrhythmic sodium channel blockers

IA- Quinidine, Procainamide, Disopyramide (prolong action potential)

IB- Lidocaine, Phenytoin, Tocainide, Mexiletine (shorten action potential)

IC- Flecainide, propafenone (slow conduction velocity

Moricizine- type 1 drug with all three characteristics
Names of Type II Anti-Arrhythmic Beta Blockers
depresses phase 4

propranolol
sotalol (also type III)
acetobutolol
esmolol
metoprolol
nadolol
atenolol
What is the difference between EAD and DAD?
Early afterdepolarization (EAD):occurs during phase 3 and is determined by calcium currents

Delayed afterdepolarization (DAD): arise when the action potential has completed and is determined by sodium and calcium channels
What are the classifications for anti-arrhythmic drugs?
type 1- sodium channel blockers
type 2- beta blockers
type 3- Potassium channel blockers
type 4- calcium channel blockers
Type I Anti-arrhythmics: Sodium-channel blockers
Type IA: Quinidine, procainamide, disopyramide (depress phase 0 and prolong action
potential) .
Type IB: Lidocaine, phenytoin, tocainide, mexiletine (depress phase 0 slightly, may shorten
action potential).
Type IC: Flecainide, propafenone (marked depression of phase 0, slow conduction velocity).
Note: moricizine is a type I drug, that shares characteristics of IA, IB and IC.
Type II Anti-arrhythmics. Beta Blockers
Propranolol, sotalol, acetobutolol, esmolol, metoprolol, nadolol, atenolol.
Sotalol may have type III effects.
Type III Anti-arrhythmics. Potassium channel blockers
(prolong phase 3 and the action
potential). Bretylium, amiodarone, sotalol.
Amiodarone has also type I, II and IV actions.
Type IV Anti-arrhythmics. Calcium channel Blockers.
Verapamil, diltiazem, bepridil.
Bepridil has additional type III effects.
effects induced by sodium channel blockers
1. Decreased excitability and automaticity
2. Decreased conduction velocity in Purkinje, myocardial and atrial cells
3. Increase refractoriness: more repolarization
What are the sodium channel blockers with addition effects?
quinidine,
procainamide and disopyramide block K-channels involved in repolarization,
prolonging action potential duration and cell refractoriness (QRS and QT
prolongation.
Type IA
It is a sodium and potassium channel blocker. It reduces excitability and automaticity, and
prolongs refractoriness.
· Prolongs QT interval (potassium channel blocking effect), and produces a modest increase
in QRS duration (sodium channel blockade of intermediate recovery time constant).
· Because of the QT prolongation it may increase the incidence of after-depolarizations
mainly at low heart rates, particularly if hypokalemia is present.
Quinidine
Type IA
Clinical use for Quinidine (Type IA)
maintenance of sinus rhythm in patients with atrial fibrillation

conversion of atrial fibrillation
Non-Cardiac side effects of Quinidine (Type IA)
Diarrhea (30-50% patients).
· Cinchonism: headache, dizziness tinnitus (improves with dose reduction).
· Immunological reactions: hepatitis, thrombocytopenia, Lupus-like syndrome, myasthenia
gravis
Why shouldn’t quinidine or disopyramide used alone to convert atrial flutter or atrial fibrillation
to normal sinus rhythm ?
Quinidine slows conduction velocity in the atrium, but the ventricular rate may increase due to its anicholinergic effects

An AV nodal blocker like Digoxin should be a pretreatment
Similar electrophysiological effects and clinical uses than quinidine.
· No alpha blocking activity.
· Strong anticholinergic activity (dry mouth, constipation, urinary retention, precipitation of
glaucoma). Its metabolite (mono-N-dealkyl-disopyramide) has stronger anticholinergic
activity than the parent drug.
· Eliminated by metabolism and urinary excretion.
Disopyramide
Similar electrophysiological effects and clinical uses than those of quinidine and
disopyramide.
· It does not have alpha blocking or anticholinergic activity.
· It is rapidly eliminated (half life: 3-4 hours), requiring repeated dosing and sustained release
formulations. Its metabolite: N-acetyl procainamide (NAPA) blocks K channels, but has
no effect on sodium channels. Parent drug prolongs QRS and QT, metabolite prolongs QT.
NAPA is eliminated by the kidney (half life 6-10 hours). Can induce Torsades de pointes.
Procainamide
Side effects of Procainamide?
leads to positive antinuclear antibodies (ANA) due to druginduced
lupus syndrome

Decreased neutrophilsÿÿÿ

>o
Common features for quinidine, disopyramide and procainamide
Very similar mechanism of action and clinical indications.
· Effects increased by hypokalemia: correct serum potassium levels prior to initiating drug
treatment.
· Caution in patients with depressed contractility and in patients receiving treatment with
cardiodepressant drugs, such as beta-blockers, CCB and other type IA drugs. It may
induce CHF.
High incidence of immunological side effects.
· Frequent monitoring of WBC, differential, platelets, ANA and LFTs.
· Excessive widening of QRS and QT prolongation (> 50%) are indicators of cardiotoxicity
(lower the dose) (must closely monitor ECG).
· Can induce Torsade de Pointes (due to QT prolongation).
· Must cardiovert or digitalize before initiating Tx for atrial fibrillation or flutter.
Differences among quinidine, disopyramide and procainamide
Quinidine- cinchonism, diarrhea, stronger anitcholinergic effects, greater alpha blocking activity

Procainamide- NAPA active antiarrhythmic metabolite, higher incidence of drug-induced lupus, shorter half-life and lower protein binding
Affects caused by Lidocain (type IB anti-arrhythmic)

side effects?
Lidocaine: is an amide local anesthetic.
· Blocks open and inactivated sodium channels (greater preference for inactivated channels).
· Main use: acute IV treatment of VT or VF.
· Decreases incidence of VF in patients with acute MI.
· Indicated for acute management of ventricular arryhthmias occurring during cardiac
manipulations, such as cardiac surgery or in relation to an acute MI.
· Not useful in atrial arryhthmias.
· Lidocaine is not given PO, because of its first pass metabolism, and the need of rapid
action in VT and VF.
· Side effects: seizures and other CNS side effects are observed when a large IV dose is
given rapidly. Proarrhythmic action. Malignant hypertermia can be produced.
Mexiletine and Tocainide (type IB)
· These agents are analogs of lidocaine with less first-pass metabolism. Their
electrophysiological actions are similar to those of lidocaine.n
Phenytoin (type 1B)
Phenytoin.
· Anticonvulsant drug (grand mal and psychomotor seizures; control of status epilepticus of
the grand mal type.).
· Like lidocaine binds to inactivated sodium channels and has a rapid recovery from block.
· Undergoes extensive first-pass metabolism and its metabolism is saturable. Small
increases in dose may produce large increases in plasma levels and toxicity (same
as propafenone). It is highly protein bound. Correct plasma levels based on albumin
concentrations.
10
· Toxicity: CNS side-efects: ataxia (incoordination), nistagmus, mental confusion,
slurred speech. Gingival hyperplasia. Rash and fever. Hematologic: bone marrow
depression, low WBC counts, low platelets, macrocytosis and megaloblastic anemia
(sensitive to folic acid).cat